1
|
Chen JK, Wiedemann J, Nguyen L, Lin Z, Tahir M, Hui CC, Plikus MV, Andersen B. IRX5 promotes DNA damage repair and activation of hair follicle stem cells. Stem Cell Reports 2023; 18:1227-1243. [PMID: 37084727 PMCID: PMC10202659 DOI: 10.1016/j.stemcr.2023.03.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 04/23/2023] Open
Abstract
The molecular mechanisms allowing hair follicles to periodically activate their stem cells (HFSCs) are incompletely characterized. Here, we identify the transcription factor IRX5 as a promoter of HFSC activation. Irx5-/- mice have delayed anagen onset, with increased DNA damage and diminished HFSC proliferation. Open chromatin regions form near cell cycle progression and DNA damage repair genes in Irx5-/- HFSCs. DNA damage repair factor BRCA1 is an IRX5 downstream target. Inhibition of FGF kinase signaling partially rescues the anagen delay in Irx5-/- mice, suggesting that the Irx5-/- HFSC quiescent phenotype is partly due to failure to suppress Fgf18 expression. Interfollicular epidermal stem cells also show decreased proliferation and increased DNA damage in Irx5-/-mice. Consistent with a role for IRX5 as a promoter of DNA damage repair, we find that IRX genes are upregulated in many cancer types and that there is a correlation between IRX5 and BRCA1 expression in breast cancer.
Collapse
Affiliation(s)
- Jefferson K Chen
- Departments of Biological Chemistry and Medicine, Division of Endocrinology, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Julie Wiedemann
- Departments of Biological Chemistry and Medicine, Division of Endocrinology, School of Medicine, University of California, Irvine, Irvine, CA, USA; Mathematical, Computational and Systems Biology (MCSB) Program, University of California, Irvine, Irvine, CA, USA
| | - Ly Nguyen
- Departments of Biological Chemistry and Medicine, Division of Endocrinology, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Zhongqi Lin
- Departments of Biological Chemistry and Medicine, Division of Endocrinology, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Mahum Tahir
- Departments of Biological Chemistry and Medicine, Division of Endocrinology, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Chi-Chung Hui
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children and Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Bogi Andersen
- Departments of Biological Chemistry and Medicine, Division of Endocrinology, School of Medicine, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
2
|
Takei Y. The digestive tract as an essential organ for water acquisition in marine teleosts: lessons from euryhaline eels. ZOOLOGICAL LETTERS 2021; 7:10. [PMID: 34154668 PMCID: PMC8215749 DOI: 10.1186/s40851-021-00175-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 04/16/2021] [Indexed: 05/17/2023]
Abstract
Adaptation to a hypertonic marine environment is one of the major topics in animal physiology research. Marine teleosts lose water osmotically from the gills and compensate for this loss by drinking surrounding seawater and absorbing water from the intestine. This situation is in contrast to that in mammals, which experience a net osmotic loss of water after drinking seawater. Water absorption in fishes is made possible by (1) removal of monovalent ions (desalinization) by the esophagus, (2) removal of divalent ions as carbonate (Mg/CaCO3) precipitates promoted by HCO3- secretion, and (3) facilitation of NaCl and water absorption from diluted seawater by the intestine using a suite of unique transporters. As a result, 70-85% of ingested seawater is absorbed during its passage through the digestive tract. Thus, the digestive tract is an essential organ for marine teleost survival in the hypertonic seawater environment. The eel is a species that has been frequently used for osmoregulation research in laboratories worldwide. The eel possesses many advantages as an experimental animal for osmoregulation studies, one of which is its outstanding euryhalinity, which enables researchers to examine changes in the structure and function of the digestive tract after direct transfer from freshwater to seawater. In recent years, the molecular mechanisms of ion and water transport across epithelial cells (the transcellular route) and through tight junctions (the paracellular route) have been elucidated for the esophagus and intestine. Thanks to the rapid progress in analytical methods for genome databases on teleosts, including the eel, the molecular identities of transporters, channels, pumps and junctional proteins have been clarified at the isoform level. As 10 y have passed since the previous reviews on this subject, it seems relevant and timely to summarize recent progress in research on the molecular mechanisms of water and ion transport in the digestive tract in eels and to compare the mechanisms with those of other teleosts and mammals from comparative and evolutionary viewpoints. We also propose future directions for this research field to achieve integrative understanding of the role of the digestive tract in adaptation to seawater with regard to pathways/mechanisms including the paracellular route, divalent ion absorption, metabolon formation and cellular trafficking of transporters. Notably, some of these have already attracted practical attention in laboratories.
Collapse
Affiliation(s)
- Yoshio Takei
- Laboratory of Physiology, Department of Marine Bioscience, Atmosphere and Ocean Research Institute, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba, 277-8564, Japan.
| |
Collapse
|
3
|
Sun X, Jiang X, Wu J, Ma R, Wu Y, Cao H, Wang Z, Liu S, Zhang J, Wu Y, Zhang Y, Feng J, Wang T. IRX5 prompts genomic instability in colorectal cancer cells. J Cell Biochem 2020; 121:4680-4689. [PMID: 32162364 DOI: 10.1002/jcb.29693] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 01/30/2020] [Indexed: 11/06/2022]
Abstract
The Iroquois homeobox gene 5 (IRX5), one of the members of the Iroquois homeobox family, has been identified to correlate with worse prognosis in many cancers, including colorectal cancer (CRC). In this study, upregulation of IRX5 revealed a great reduction in the proliferation of CRC colorectal cancer cell line SW480 and DLD-1, which was accompanied by G1/S arrest, increased expression in cyclin E1, P21, and P53 and a decrease in cyclin A2, B1, and D1. Furthermore, IRX5-mediated an increase expression of RH2A protein, the biomarker of DNA damage. Consequently, the SA-β-gal level is higher in IRX5-overexpression cells compared to control ones, which showed elevated DNA damage triggered cellular senescence. Recapitulating the above findings, IRX5 exhibited higher levels of genomic instability. IRX5 may be a perspective target for cancer therapy and it deserves further investigation.
Collapse
Affiliation(s)
- Xun Sun
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Xinying Jiang
- Department of Cell Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jianzhong Wu
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Rong Ma
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Yiqi Wu
- Department of Cell Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Haixia Cao
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Zhuo Wang
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Siwen Liu
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Junying Zhang
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Yang Wu
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Yuan Zhang
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Jifeng Feng
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Ting Wang
- Department of Cell Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
4
|
Bjune JI, Dyer L, Røsland GV, Tronstad KJ, Njølstad PR, Sagen JV, Dankel SN, Mellgren G. The homeobox factor Irx3 maintains adipogenic identity. Metabolism 2020; 103:154014. [PMID: 31751577 DOI: 10.1016/j.metabol.2019.154014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 10/22/2019] [Accepted: 11/13/2019] [Indexed: 01/13/2023]
Abstract
BACKGROUND Inhibition of Irx3 and Irx5 has been shown to reduce body weight and white adipose tissue (WAT) mass through cell-autonomous and sympathetic-induced increases in adipocyte beiging and thermogenesis in mice and humans. However, the underlying mechanisms of the Irx control over beiging are still largely unknown, as illustrated by recent reports showing divergent effects of Irx3 on adipocyte metabolism and function. Here, we investigated the role of Irx3 in controlling beige preadipocyte function and differentiation. METHODS Stable knock out of Irx3 in ME3 mouse preadipocytes capable of beiging was performed using a CRISPR-Cas9 system, and the effect on cell differentiation was assessed by qPCR, RNA-seq, Oil-red-O lipid staining and Alcian Blue staining of proteoglycans. Changes in cell identities were validated using cell type enrichment analysis from RNA-seq data. Proliferation and cell cycle progression in undifferentiated cells were measured by WST-1 and flow cytometry, reactive oxygen species (ROS) generation was determined by fluorescence spectrometry and mitochondrial respiration was investigated by Seahorse assay. RESULTS Irx3 was found to be essential for the identity, function and adipogenic differentiation of beige adipocyte precursors. Irx3-KO impaired proliferation, ROS generation and mitochondrial respiration in the preadipocytes. We further observed profound changes in numerous genes during both early and late stages of adipogenic differentiation, including genes important for adipocyte differentiation, cell cycle progression, oxidative phosphorylation (OXPHOS) and morphogenesis. Irx3-KO cells failed to accumulate lipids following adipogenic stimuli, and cell enrichment analysis revealed a loss of preadipocyte identity and a gain of chondrocyte-like identity in Irx3-KO cells during early differentiation. Finally, unlike the control cells, the Irx3-KO cells readily responded to chondrogenic stimuli. CONCLUSIONS Irx3 is required for preadipocyte identity and differentiation capacity. Our findings suggest that, while inhibition of Irx3 may be beneficial during later developmental stages to modulate adipogenesis in the beige direction, constitutive and complete absence of Irx3 in the embryonic fibroblast stage leads to detrimental loss of adipogenic differentiation capacity.
Collapse
Affiliation(s)
- Jan-Inge Bjune
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway; Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway; Hormone Laboratory, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Laurence Dyer
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway; Hormone Laboratory, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Gro V Røsland
- Department of Biomedicine, University of Bergen, N-5020 Bergen, Norway
| | - Karl Johan Tronstad
- Department of Biomedicine, University of Bergen, N-5020 Bergen, Norway; Department of Oncology and Medical Physics, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Pål R Njølstad
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway; Department of Pediatrics and Adolescents, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Jørn V Sagen
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway; Hormone Laboratory, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Simon N Dankel
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway; Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway; Hormone Laboratory, Haukeland University Hospital, N-5021 Bergen, Norway.
| | - Gunnar Mellgren
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway; Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway; Hormone Laboratory, Haukeland University Hospital, N-5021 Bergen, Norway.
| |
Collapse
|
5
|
Kalbuaji B, Taguchi YH, Konagaya A. Discovery of a Robust Gene Regulatory Network with a Complex Transcription Factor Network on Organ Cancer Cell-line RNA Sequence Data. CHEM-BIO INFORMATICS JOURNAL 2019. [DOI: 10.1273/cbij.19.32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
6
|
Zhu Q, Wu Y, Yang M, Wang Z, Zhang H, Jiang X, Chen M, Jin T, Wang T. IRX5 promotes colorectal cancer metastasis by negatively regulating the core components of the RHOA pathway. Mol Carcinog 2019; 58:2065-2076. [PMID: 31432570 DOI: 10.1002/mc.23098] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 07/27/2019] [Accepted: 07/31/2019] [Indexed: 01/20/2023]
Abstract
Colorectal cancer (CRC) is one of the most common malignant tumors worldwide. As tumor metastasis is the leading cause of death in patients with CRC, it is important to elucidate the molecular mechanisms that drive CRC metastasis. Studies have shown a close relationship between Iroquois homeobox (IRX) family genes and multiple cancers, while the mechanism by which IRX5 promotes CRC metastasis is unclear. Therefore, we focused on the involvement of IRX5 in CRC metastasis. In this study, analyses of clinical data indicated that the expression of IRX5 was coincided with metastatic colorectal tumors tissues and was negatively correlated with the overall survival of patients with CRC. Functional analysis showed that IRX5 promoted the migration and invasion of CRC cells, accompanied by a large number of cellular protrusions. IRX5-overexpressing cells were more likely to form metastatic tumors in nude mice. Further analysis demonstrated that the core components of the RHOA/ROCK1/LIMK1 pathway were significantly inhibited in IRX5-overexpressing cells. Overexpression of LIMK1 effectively reversed the enhanced cellular motility caused by IRX5 overexpression. Moreover, we found that high levels of IRX5 in intestinal tissues were correlated with the inflammatory response. IRX5 was significantly increased in azoxymethane/dextran sodium sulfate intestinal tissue of mice and IRX5-overexpressing may also enhance chemokines CXCL1 and CXCL8. In summary, our findings suggested that IRX5 promoted CRC metastasis by inhibiting the RHOA-ROCK1-LIMK1 axis, which correlates with a poor prognosis.
Collapse
Affiliation(s)
- Qiangqiang Zhu
- Department of Cell Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yiqi Wu
- Department of Cell Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Mengli Yang
- Department of Cell Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhen Wang
- Department of Pathology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hailing Zhang
- Department of Cell Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xinying Jiang
- Department of Cell Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Meng Chen
- Department of Molecular, Cellular and Biomedical Sciences, Medical Laboratory Science Program, College of Life Sciences and Agriculture, The University of New Hampshire, Durham, New Hampshire
| | - Tianyu Jin
- Department of Clinic School, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ting Wang
- Department of Cell Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
7
|
Mitchell D, Chintala S, Fetcko K, Henriquez M, Tewari BN, Ahmed A, Bentley RT, Dey M. Common Molecular Alterations in Canine Oligodendroglioma and Human Malignant Gliomas and Potential Novel Therapeutic Targets. Front Oncol 2019; 9:780. [PMID: 31475119 PMCID: PMC6702544 DOI: 10.3389/fonc.2019.00780] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 07/31/2019] [Indexed: 01/05/2023] Open
Abstract
Spontaneous canine (Canis lupus) oligodendroglioma (ODG) holds tremendous potential as an immunocompetent large animal model of human malignant gliomas (MG). However, the feasibility of utilizing this model in pre-clinical studies depends on a thorough understanding of the similarities and differences of the molecular pathways associated with gliomas between the two species. We have previously shown that canine ODG has an immune landscape and expression pattern of commonly described oncogenes similar to that of human MG. In the current study, we performed a comprehensive analysis of canine ODG RNAseq data from 4 dogs with ODG and 2 normal controls to identify highly dysregulated genes in canine tumors. We then evaluated the expression of these genes in human MG using Xena Browser, a publicly available database. STRING-database inquiry was used in order to determine the suggested protein associations of these differentially expressed genes as well as the dysregulated pathways commonly enriched by the protein products of these genes in both canine ODG and human MG. Our results revealed that 3,712 (23%) of the 15,895 differentially expressed genes demonstrated significant up- or downregulation (log2-fold change > 2.0). Of the 3,712 altered genes, ~50% were upregulated (n = 1858) and ~50% were downregulated (n = 1854). Most of these genes were also found to have altered expression in human MG. Protein association and pathway analysis revealed common pathways enriched by members of the up- and downregulated gene categories in both species. In summary, we demonstrate that a similar pattern of gene dysregulation characterizes both human MG and canine ODG and provide additional support for the use of the canine model in order to therapeutically target these common genes. The results of such therapeutic targeting in the canine model can serve to more accurately predict the efficacy of anti-glioma therapies in human patients.
Collapse
Affiliation(s)
- Dana Mitchell
- Department of Neurosurgery, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Sreenivasulu Chintala
- Department of Neurosurgery, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Kaleigh Fetcko
- Department of Neurosurgery, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Mario Henriquez
- Department of Neurosurgery, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Brij N Tewari
- Department of Neurosurgery, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Atique Ahmed
- Department of Neurological Surgery, Northwestern University, Chicago, IL, United States
| | - R Timothy Bentley
- Department of Veterinary Clinical Sciences, Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - Mahua Dey
- Department of Neurosurgery, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
8
|
Takei Y, Wong MKS, Ando M. Molecular mechanisms for intestinal HCO3− secretion and its regulation by guanylin in seawater-acclimated eels. J Exp Biol 2019; 222:jeb.203539. [DOI: 10.1242/jeb.203539] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 06/03/2019] [Indexed: 01/25/2023]
Abstract
The intestine of marine teleosts secretes HCO3− into the lumen and precipitates Ca2+ and Mg2+ in the imbibed seawater as carbonates to decrease luminal fluid osmolality and facilitate water absorption. However, hormonal regulation of HCO3−secretion is largely unknown. Here, mucosally-added guanylin (GN) increased HCO3− secretion, measured by pH-stat, across isolated seawater-acclimated eel intestine bathed in saline at pH 7.4 (5% CO2). The effect of GN on HCO3− secretion was slower than that on the short-circuit current, and the time-course of the GN effect was similar to that of bumetanide. Mucosal bumetanide and serosal 4,4’-dinitrostilbene-2,2’-disulfonic acid (DNDS) inhibited the GN effect, suggesting an involvement of apical Na+-K+-2Cl− cotransporter (NKCC2) and basolateral Cl−/HCO3− exchanger (AE)/Na+-HCO3− cotransporter (NBC) in the GN effect. As mucosal DNDS failed to inhibit the GN effect, apical DNDS-sensitive AE may not be involved. To identify molecular species of transporters involved in the GN effect, we performed RNA-seq analyses followed by quantitative real-time PCR after transfer of eels to seawater. Among the genes upregulated after seawater transfer, AE genes, draa, b, and pat1a, c, on the apical membrane, and NBC genes, nbce1a, n1, n2a, and a AE gene, sat-1, on the basolateral membrane were candidates involved in HCO3− secretion. Judging from the slow effect of GN, we suggest that GN inhibits NKCC2b on the apical membrane and decreases cytosolic Cl− and Na+, which then activates apical DNDS-insensitive DRAs and basolateral DNDS-sensitive NBCs to enhance transcellular HCO3− flux across the intestinal epithelia of seawater-acclimated eels.
Collapse
Affiliation(s)
- Yoshio Takei
- Laboratory of Physiology, Atmosphere and Ocean Research Institute, the University of Tokyo, Kashiwa, Chiba 277-8564, Japan
| | - Marty K. S. Wong
- Laboratory of Physiology, Atmosphere and Ocean Research Institute, the University of Tokyo, Kashiwa, Chiba 277-8564, Japan
| | - Masaaki Ando
- Laboratory of Physiology, Atmosphere and Ocean Research Institute, the University of Tokyo, Kashiwa, Chiba 277-8564, Japan
| |
Collapse
|
9
|
Huang L, Song F, Sun H, Zhang L, Huang C. IRX5 promotes NF-κB signalling to increase proliferation, migration and invasion via OPN in tongue squamous cell carcinoma. J Cell Mol Med 2018; 22:3899-3910. [PMID: 29761910 PMCID: PMC6050492 DOI: 10.1111/jcmm.13664] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Accepted: 03/26/2018] [Indexed: 12/11/2022] Open
Abstract
Iroquois homeobox gene 5 (Irx5) is a highly conserved member of the Iroquois homeobox gene family. Members of this family play distinct and overlapping roles in normal embryonic cell patterning and development of malignancies. In this study, we observed that IRX5 was abnormally abundant in tongue squamous cell carcinoma (TSCC) tissues and cell lines. We used gain‐ and loss‐of‐function methods to overexpress and knockdown IRX5 expression in the TSCC cell line CAL27. Our results elucidated that elevated levels of IRX5 promoted proliferation, migration and invasion of TSCC cells, whereas stable or transient knockdown of IRX5 expression suppressed TSCC cell proliferation, migration and invasion. As a transcription factor, IRX5 performed this function by targeting osteopontin (OPN) promoter and activating the NF‐κB pathway. Finally, studies in xenograft tumour model showed that IRX5 significantly enhanced OPN expression and promoted tumour growth. Taken together, our study elucidates a promotive effect of IRX5 in TSCC through the connection with OPN. These findings reveal the new molecular mechanism of TSCC, which may potentiate its use as a novel molecular therapy target for TSCC.
Collapse
Affiliation(s)
- Liyuan Huang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBM), School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Fangfang Song
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBM), School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Hualing Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBM), School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Lu Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBM), School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Cui Huang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBM), School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
10
|
Wang Y, Xu P, Qiu L, Zhang M, Huang Y, Zheng JC. CXCR7 Participates in CXCL12-mediated Cell Cycle and Proliferation Regulation in Mouse Neural Progenitor Cells. Curr Mol Med 2017; 16:738-746. [PMID: 27573194 PMCID: PMC5345320 DOI: 10.2174/1566524016666160829153453] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 08/23/2016] [Accepted: 08/24/2016] [Indexed: 12/30/2022]
Abstract
Background: Cell cycle regulation of neural progenitor cells (NPCs) is an essential process for neurogenesis, neural development, and repair after brain trauma. Stromal cell-derived factor-1 (SDF-1, CXCL12) and its receptors CXCR4 and CXCR7 are well known in regulating the migration and survival of NPCs. The effects of CXCL12 on NPCs proliferation, cell cycle regulation, and their associated signaling pathways remain unclear. Cyclin D1 is a protein required for progression through the G1 phase of the cell cycle and a known downstream target of β-catenin. Therefore, cyclin D1 plays critical roles of cell cycle regulation, proliferation, and survival in NPCs. Methods: Primary mouse NPCs (mNPCs) were derived from brain tissues of wild-type, Cxcr4 knockout, or Cxcr7 knockout mice at mouse embryonic day 13.5 (E13.5). Flow cytometry was used to perform cell cycle analysis by quantitation of DNA content. Real-time PCR and Western blot were used to evaluate mRNA and protein expressions, respectively. Ki67 immunostaining and TUNEL assay were used to assess the proliferation and survival of mNPCs, respectively. Results: CXCL12 pretreatment led to the shortening of G0/G1 phase and lengthening of S phase, suggesting that CXCL12 regulates cell cycle progression in mNPCs. Consistently, CXCL12 treatment increased the expression of CyclinD1 and β-catenin, and promoted proliferation and survival of mNPCs. Cxcr7 knockout of mNPCs blocked CXCL12-mediated mNPCs proliferation, whereas Cxcr4 knockout mNPC did not significantly effect CXCL12- mediated mNPCs proliferation. Conclusion: CXCR7 plays an important role in CXCL12-mediated mNPC cell cycle regulation and proliferation.
Collapse
Affiliation(s)
| | | | | | | | - Y Huang
- Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai 200072, China; 985930 University of Nebraska Medical Center, Omaha, NE 68198-5930, USA.
| | | |
Collapse
|